scispace - formally typeset
Search or ask a question

Showing papers by "Weidong Han published in 2023"


Journal ArticleDOI
TL;DR: Wang et al. as mentioned in this paper performed a retrospective study on patients with unresectable biliary tract cancer who received a combination of programmed death 1 inhibitor and antiangiogenic agent between March 26, 2019 and November 1, 2021.
Abstract: Immunotherapy and antiangiogenic therapy have shown promising clinical activity in patients with advanced biliary tract cancer (BTC) in clinical trials. As the combination of these two treatments for BTC is not well studied in the real world, the present study retrospectively analyzed the clinical outcomes of patients with unresectable BTC who received immunotherapy-antiangiogenesis combination therapy in a real-world setting. A three-center, retrospective study was performed on patients with unresectable BTC who received a combination of programmed death 1 inhibitor and antiangiogenic agent between March 26, 2019 and November 1, 2021 in China. In total, 68 patients were enrolled in the cohort. The objective response rate and disease control rate were 13.2 and 75.0%, respectively. The median time to progression, progression-free survival and overall survival were 8.2, 5.5 and 10.7 months, respectively. Adverse events of all grades occurred in 58 patients (85.3%). In conclusion, the present study demonstrated that immunotherapy-antiangiogenesis combination therapy may be considered a therapeutic option for patients with unresectable BTC. Further prospective investigations are needed.

Journal ArticleDOI
TL;DR: A first-in-human phase Ia dose escalation study of Ametumumab in patients with advanced solid malignancies was conducted in this article , where patients received six ascending dosages ranging from 75 to 750 mg/m2.
Abstract: Background: Epidermal growth factor receptor (EGFR) is a well-known target for cancer treatment. However, the authorized anti-EGFR monoclonal antibodies generally cause several toxic effects, especially severe cutaneous toxicities as well as infusion reactions, and the clinical indications are limited. Here we developed Ametumumab, a fully human recombinant anti-EGFR monoclonal antibody. Objectives: To assess the safety, tolerability, pharmacokinetics (PK), and immunogenicity of Ametumumab. Design: A first-in-human phase Ia dose escalation study of Ametumumab in patients with advanced solid malignancies. Methods: An open-label, first-in-human dose escalation study was done in 22 patients with advanced malignancies who received six ascending dosages ranging from 75 to 750 mg/m2. Following a single dosage and a 28-day dose-limiting toxicity (DLT) monitoring period, patients were given repeated doses weekly. Blood samples were taken to determine the PK parameters of Ametumumab and anti-drug antibody concentrations. Every 8 weeks, radiographic tumor evaluations were conducted. Results: In this trial, no DLT was observed, and the maximum tolerated dose was not reached at doses up to 750 mg/m2. There were no severe adverse events but mild and moderate adverse effects, such as headache, proteinuria, and rash. Single-dose PK results demonstrated a straightforward linear relationship with dosage escalation. The medication concentrations accumulated and attained steady-state after four rounds of injections. It was calculated that 10 patients with disease control would be observed in the 22 evaluable patients. The disease control rate was 45.5%. Conclusion: The Ametumumab was well tolerated and safe in patients with advanced solid malignancies, exhibiting minimal immunogenicity, a long half-life, high levels of drug exposure in the blood, and preliminary effectiveness. Registration: The trial was registered with CTR20170343 on 10 April 2017, The China Center for Drug Evaluation.

Journal ArticleDOI
TL;DR: In this article , GFH018, a small molecule inhibiting TGF-βRI kinase, blocks TGFβ signaling transduction thus downregulates its pathway, and demonstrated significant growth inhibitory effects on several in vivo tumor models as monotherapy.
Abstract: e15117 Background: TGF-β signaling pathway activation inhibits anti-tumor response, promoting tumor progression and metastasis. GFH018, a small molecule inhibiting TGF-βRI kinase, blocks TGF-β signaling transduction thus downregulates its pathway. GFH018 had significant growth inhibitory effects on several in vivo tumor models as monotherapy and demonstrated synergistic effects combined with anti-PD-1 antibody. This phase I study assessed safety, PK, and preliminary efficacy of GFH018 in pts with advanced solid tumors. Here we report the final results from the completed study. Methods: The study includes a modified 3 + 3 dose escalation and an expansion part. Adult pts with advanced solid tumors failed to standard treatments were enrolled. The starting dose was 5 mg and up to 8 cohorts were planned. In the escalation part, pts were administrated with GFH018 BID (14d-on/14d-off) two days after receiving single dose orally on C1D1. The subsequent cycle was 28 days. The safety of 85 mg BID 7d-on/7d-off was also explored. AEs were graded per NCI-CTCAE v5.0. PK was analyzed using a non-compartmental method. Efficacy was evaluated per RECIST 1.1. Blood samples were collected for biomarker analysis. Results: From Aug 1, 2019 to Aug 11, 2022, fifty pts (14d-on/14d-off: 5 mg [n = 4], 10 mg [n = 3], 20mg [n = 4], 30 mg [n = 7], 40 mg [n = 4], 50 mg [n = 4], 65 mg [n = 6] and 85 mg [n = 12]; 7d-on/7d-off: 85 mg [n = 6]) were enrolled. Thirty-seven pts (74.0%) had ≥3 prior systemic treatment lines. No DLT was observed, and the MTD was not reached. No significant cardiotoxicities or bleeding events were reported. Forty-three pts (86.0%) had at least one treatment related AE (TRAE), and 3 (6.0%) experienced ≥ G3 TRAEs. The most common TRAEs (all G/≥G3) were proteinuria (26.0%/2.0%), AST increased (18.0%/0), anemia (14.0%/2.0%), ALT increased (12.0%/0), GGT increased, ALP increased, and LDH increased (all 10.0%/0). Common TRAEs (≥2 pts) for 7d-on/7d-off was lymphocyte decreased (2/6) with no ≥ G3 reported. No significant differences were observed between the two regimens in safety. GFH018 was absorbed rapidly with median Tmax of 0.50 ~0.96h and displayed a linear pharmacokinetic behavior with T1/2 of 4.08~10.92 h (5~85 mg). Nine pts achieved stable disease (SD), among whom a pt with thymic carcinoma receiving 65 mg achieved tumor shrinkage (maximum target lesion decreased by 18.4%). Results for 85 mg BID, 14d-on/14d-off showed a DCR of 25.0%. The RP2D was 85 mg BID, 14d-on/14d-off. Serum TGF-β1 level was not associated with clinical responses. Available pharmacodynamic data of SMAD2 phosphorylation levels in peripheral blood mononuclear cells (PBMCs) were not reliable due to assay issues. Conclusions: GFH018 shows a favorable safety profile and preliminary anti-tumor activity. The clinical studies of GFH018 in combination with Toripalimab and with Toripalimab + concurrent chemoradiotherapy are ongoing. Clinical trial information: NCT05051241 .

Journal ArticleDOI
TL;DR: In this paper , a 52-year-old female was admitted to hospital for dull abdominal pain, with peripancreatic lesions below the caudate lobe of the liver and posterior peritoneal lymph nodes enlargement.
Abstract: Background Cancer of unknown primary (CUP) is a malignant and aggressive tumor whose primary origin is still unknown despite thorough evaluation. CUP can be life-threatening with a median overall survival of less than 1 year based on empirical chemotherapy. Gene detection technology advances the driver gene detection of malignant tumors and the appropriate precise therapy. Immunotherapy has ushered in a new era in cancer therapy, changing the way advanced tumors, including CUP, are treated. Combined with comprehensive clinical and pathological investigations, molecular analysis of the original tissue and detection of potential driver mutations may provide therapeutic recommendations for CUP. Case presentation A 52-year-old female was admitted to hospital for dull abdominal pain, with peripancreatic lesions below the caudate lobe of the liver and posterior peritoneal lymph nodes enlargement. Conventional biopsy under endoscopic ultrasonography and laparoscopic biopsy both revealed poorly differentiated adenocarcinoma based on immunohistochemical series. To help identify tumor origin and molecular characteristics, 90-gene expression assay, tumor gene expression profiling with Next-generation sequencing (NGS) method and Immunohistochemical expression of PD-L1 were employed. Although no gastroesophageal lesions discovered by gastroenteroscopy, the 90-gene expression assay yielded a similarity score and prompted the most likely primary site was gastric/esophagus cancer. NGS revealed high TMB (19.3mutations/Mb) but no druggable driver genes identified. The Dako PD-L1 22C3 assay IHC assay for PD-L1 expression revealed a tumor proportion score (TPS) of 35%. Given the presence of negative predictive biomarkers for immunotherapy, including adenomatous polyposis coli (APC) c.646C>T mutation at exon 7 and Janus kinase 1(JAK1), the patient received immunochemotherapy instead of immunotherapy alone. She was successfully treated with nivolumab plus carboplatin and albumin-bound nanoparticle paclitaxel for six cycles and nivolumab maintenance, which achieved a complete response (CR) maintained for 2 years without severe adverse events. Conclusions This case highlights the value of multidisciplinary diagnosis and individual precision treatment in CUP. Further investigation is needed as an individualized treatment approach combining immunotherapy and chemotherapy based on tumor molecular characteristics and immunotherapy predictors is expected to improve the outcome of CUP therapy.

Journal ArticleDOI
TL;DR: In this paper , the authors reported the first case of concomitant ALK rearrangement and SMARCA4 deficiency in a nonsmoking female with thoracic cancer.
Abstract: Recently, SMARCA4-deficient undifferentiated tumor (SMARCA4-UT) has emerged as a distinct subset of lung cancer. Previous studies have suggested that SMARCA4-UT is often associated with smoking-related mutations, such as KRAS and STK11, rather than EGFR or ALK alterations. Nevertheless, no specific precision therapy has been identified for SMARCA4-UT. Here, we report the first case of concomitant ALK rearrangement and SMARCA4 (BRG1) deficiency in a nonsmoking female with thoracic cancer. Alectinib was given as the first-line therapy, and the patient achieved a remarkable complete response. Our case highlights the significance of ALK rearrangement identification for the precise therapeutic potential of SMARCA4-UT.